Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
J Med Chem ; 65(2): 1552-1566, 2022 01 27.
Article in English | MEDLINE | ID: mdl-34958586

ABSTRACT

Dabrafenib is an anticancer drug currently used in the clinics, alone or in combination. However, dabrafenib was recently shown to potently activate the human nuclear receptor pregnane X receptor (PXR). PXR activation increases the clearance of various chemicals and drugs, including dabrafenib itself. It may also enhance cell proliferation and tumor aggressiveness. Therefore, there is a need for rational design of a potent protein kinase B-Raf inhibitor devoid of binding to the secondary target PXR and resisting rapid metabolism. By determining the crystal structure of dabrafenib bound to PXR and analyzing its mode of binding to both PXR and its primary target, B-Raf-V600E, we were able to derive new compounds with nanomolar activity against B-Raf and no detectable affinity for PXR. The crystal structure of B-Raf in complex with our lead compound revealed a subdomain swapping of the activation loop with potentially important functional implications for a prolonged inhibition of B-Raf-V600E.


Subject(s)
Cell Proliferation , Drug Design , Imidazoles/pharmacology , Melanoma/drug therapy , Oximes/pharmacology , Pregnane X Receptor/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Crystallography, X-Ray , Humans , Imidazoles/chemistry , Melanoma/pathology , Molecular Docking Simulation , Oximes/chemistry , Protein Binding , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
2.
BMC Biol ; 19(1): 248, 2021 11 20.
Article in English | MEDLINE | ID: mdl-34801011

ABSTRACT

BACKGROUND: Lipid species are accurately distributed in the eukaryotic cell so that organelle and plasma membranes have an adequate lipid composition to support numerous cellular functions. In the plasma membrane, a precise regulation of the level of lipids such as phosphatidylserine, PI(4)P, and PI(4,5)P2, is critical for maintaining the signaling competence of the cell. Several lipid transfer proteins of the ORP/Osh family contribute to this fine-tuning by delivering PS, synthesized in the endoplasmic reticulum, to the plasma membrane in exchange for PI(4)P. To get insights into the role of these PS/PI(4)P exchangers in regulating plasma membrane features, we question how they selectively recognize and transfer lipid ligands with different acyl chains, whether these proteins exchange PS exclusively for PI(4)P or additionally for PI(4,5)P2, and how sterol abundance in the plasma membrane impacts their activity. RESULTS: We measured in vitro how the yeast Osh6p and human ORP8 transported PS and PI(4)P subspecies of diverse length and unsaturation degree between membranes by fluorescence-based assays. We established that the exchange activity of Osh6p and ORP8 strongly depends on whether these ligands are saturated or not, and is high with representative cellular PS and PI(4)P subspecies. Unexpectedly, we found that the speed at which these proteins individually transfer lipid ligands between membranes is inversely related to their affinity for them and that high-affinity ligands must be exchanged to be transferred more rapidly. Next we determined that Osh6p and ORP8 cannot use PI(4,5)P2 for exchange processes, because it is a low-affinity ligand, and do not transfer more PS into sterol-rich membranes. CONCLUSIONS: Our study provides new insights into PS/PI(4)P exchangers by indicating the degree to which they can regulate the acyl chain composition of the PM, and how they control PM phosphoinositide levels. Moreover, we establish general rules on how the activity of lipid transfer proteins relates to their affinity for ligands.


Subject(s)
Phosphatidylserines , Saccharomyces cerevisiae , Cell Membrane , Endoplasmic Reticulum , Humans , Ligands , Sterols
3.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Article in English | MEDLINE | ID: mdl-33361153

ABSTRACT

Humans are chronically exposed to mixtures of xenobiotics referred to as endocrine-disrupting chemicals (EDCs). A vast body of literature links exposure to these chemicals with increased incidences of reproductive, metabolic, or neurological disorders. Moreover, recent data demonstrate that, when used in combination, chemicals have outcomes that cannot be predicted from their individual behavior. In its heterodimeric form with the retinoid X receptor (RXR), the pregnane X receptor (PXR) plays an essential role in controlling the mammalian xenobiotic response and mediates both beneficial and detrimental effects. Our previous work shed light on a mechanism by which a binary mixture of xenobiotics activates PXR in a synergistic fashion. Structural analysis revealed that mutual stabilization of the compounds within the ligand-binding pocket of PXR accounts for the enhancement of their binding affinity. In order to identify and characterize additional active mixtures, we combined a set of cell-based, biophysical, structural, and in vivo approaches. Our study reveals features that confirm the binding promiscuity of this receptor and its ability to accommodate bipartite ligands. We reveal previously unidentified binding mechanisms involving dynamic structural transitions and covalent coupling and report four binary mixtures eliciting graded synergistic activities. Last, we demonstrate that the robust activity obtained with two synergizing PXR ligands can be enhanced further in the presence of RXR environmental ligands. Our study reveals insights as to how low-dose EDC mixtures may alter physiology through interaction with RXR-PXR and potentially several other nuclear receptor heterodimers.


Subject(s)
Pregnane X Receptor/chemistry , Retinoid X Receptors/chemistry , Xenobiotics , Animals , Cell Line , Crystallography, X-Ray , Dimerization , Fluorescence Polarization , Gene Expression Regulation , Humans , Ligands , Luciferases/genetics , Luciferases/metabolism , Models, Chemical , Pregnane X Receptor/metabolism , Retinoid X Receptors/metabolism , Xenobiotics/chemistry , Xenobiotics/metabolism , Xenobiotics/pharmacology , Xenopus
4.
Cell Mol Life Sci ; 76(23): 4769-4781, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31127318

ABSTRACT

The estrogen-related receptor γ (ERRγ, NR3B3) is a constitutively active nuclear receptor which has been proposed to act as a mediator of the low-dose effects of a number of environmental endocrine-disrupting chemicals (EDCs) such as the xenoestrogen bisphenol-A (BPA). To better characterize the ability of exogenous compounds to bind and activate ERRγ, we used a combination of cell-based, biochemical, structural and computational approaches. A purposely created stable cell line allowed for the determination of the EC50s for over 30 environmental ERRγ ligands, including previously unknown ones. Interestingly, affinity constants (Kds) of the most potent compounds measured by isothermal titration calorimetry were in the 50-500 nM range, in agreement with their receptor activation potencies. Crystallographic analysis of the interaction between the ERRγ ligand-binding domain (LBD) and compounds of the bisphenol, alkylphenol and naphthol families revealed a partially shared binding mode and minimal alterations of the receptor conformation upon ligand binding. Further biophysical characterizations coupled to molecular dynamics simulations suggested a mechanism through which ERRγ ligands would exhibit their agonistic properties by preserving the transcriptionally active form of the receptor while rigidifying some loop regions with associated functions. This unique mechanism contrasts with the classical one involving a ligand-induced repositioning and stabilization of the C-terminal activation helix H12.


Subject(s)
Endocrine Disruptors/chemistry , Receptors, Estrogen/metabolism , Benzhydryl Compounds/chemistry , Benzhydryl Compounds/metabolism , Benzhydryl Compounds/pharmacology , Binding Sites , Cell Line, Tumor , Crystallography, X-Ray , Endocrine Disruptors/metabolism , Endocrine Disruptors/pharmacology , Humans , Ligands , Molecular Dynamics Simulation , Phenols/chemistry , Phenols/metabolism , Phenols/pharmacology , Protein Binding , Protein Structure, Tertiary , Receptors, Estrogen/chemistry , Receptors, Estrogen/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Thermodynamics , Transcriptional Activation/drug effects
5.
C R Biol ; 340(9-10): 414-420, 2017.
Article in English | MEDLINE | ID: mdl-29126514

ABSTRACT

Endocrine-disrupting chemicals (EDCs) represent a broad class of exogenous substances that cause adverse effects in the endocrine system mainly by interacting with nuclear hormone receptors (NRs). Humans are generally exposed to low doses of pollutants, and current researches aim at deciphering the mechanisms accounting for the health impact of EDCs at environmental concentrations. Our correlative analysis of structural, interaction and cell-based data has revealed a variety of, sometimes unexpected, binding modes, reflecting a wide range of EDC affinities and specificities. Here, we present a few representative examples to illustrate various means by which EDCs achieve high-affinity binding to NRs. These examples include the binding of the mycoestrogen α-zearalanol to estrogen receptors, the covalent interaction of organotins with the retinoid X- and peroxisome proliferator-activated receptors, and the cooperative binding of two chemicals to the pregnane X receptor. We also discuss some hypotheses that could further explain low-concentration effects of EDCs with weaker affinity towards NRs.


Subject(s)
Endocrine Disruptors/toxicity , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Humans
7.
Nat Commun ; 6: 8089, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26333997

ABSTRACT

Humans are chronically exposed to multiple exogenous substances, including environmental pollutants, drugs and dietary components. Many of these compounds are suspected to impact human health, and their combination in complex mixtures could exacerbate their harmful effects. Here we demonstrate that a pharmaceutical oestrogen and a persistent organochlorine pesticide, both exhibiting low efficacy when studied separately, cooperatively bind to the pregnane X receptor, leading to synergistic activation. Biophysical analysis shows that each ligand enhances the binding affinity of the other, so the binary mixture induces a substantial biological response at doses at which each chemical individually is inactive. High-resolution crystal structures reveal the structural basis for the observed cooperativity. Our results suggest that the formation of 'supramolecular ligands' within the ligand-binding pocket of nuclear receptors contributes to the synergistic toxic effect of chemical mixtures, which may have broad implications for the fields of endocrine disruption, toxicology and chemical risk assessment.


Subject(s)
Estrogens/pharmacology , Ethinyl Estradiol/pharmacology , Hydrocarbons, Chlorinated/pharmacology , Insecticides/pharmacology , Receptors, Steroid/drug effects , Blotting, Western , Cell Line , Cell Line, Tumor , Crystallization , Crystallography, X-Ray , Cytochrome P-450 CYP3A/drug effects , Cytochrome P-450 CYP3A/metabolism , Drug Synergism , Environmental Pollutants/chemistry , Environmental Pollutants/pharmacology , Estrogens/chemistry , Ethinyl Estradiol/chemistry , Fluorescence Polarization , Hep G2 Cells , Hepatocytes , Humans , Hydrocarbons, Chlorinated/chemistry , Insecticides/chemistry , Mass Spectrometry , Pregnane X Receptor , Real-Time Polymerase Chain Reaction , Receptors, Steroid/chemistry , Retinoid X Receptors/drug effects , Retinoid X Receptors/metabolism , Reverse Transcriptase Polymerase Chain Reaction
8.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 8): 1777-87, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26249358

ABSTRACT

X-ray crystallography is an established technique for ligand screening in fragment-based drug-design projects, but the required manual handling steps - soaking crystals with ligand and the subsequent harvesting - are tedious and limit the throughput of the process. Here, an alternative approach is reported: crystallization plates are pre-coated with potential binders prior to protein crystallization and X-ray diffraction is performed directly 'in situ' (or in-plate). Its performance is demonstrated on distinct and relevant therapeutic targets currently being studied for ligand screening by X-ray crystallography using either a bending-magnet beamline or a rotating-anode generator. The possibility of using DMSO stock solutions of the ligands to be coated opens up a route to screening most chemical libraries.


Subject(s)
Crystallization/methods , Crystallography, X-Ray/methods , Drug Discovery/methods , Proteins/chemistry , Animals , Chickens , Peptidyl-Prolyl Isomerase F , Cyclophilins/chemistry , Cyclophilins/metabolism , Humans , Ligands , Mitogen-Activated Protein Kinase 1/chemistry , Mitogen-Activated Protein Kinase 1/metabolism , Muramidase/chemistry , Muramidase/metabolism , PPAR gamma/chemistry , PPAR gamma/metabolism , Proteins/metabolism , Rats
9.
Science ; 349(6246): 432-6, 2015 Jul 24.
Article in English | MEDLINE | ID: mdl-26206936

ABSTRACT

In eukaryotic cells, phosphatidylserine (PS) is synthesized in the endoplasmic reticulum (ER) but is highly enriched in the plasma membrane (PM), where it contributes negative charge and to specific recruitment of signaling proteins. This distribution relies on transport mechanisms whose nature remains elusive. Here, we found that the PS transporter Osh6p extracted phosphatidylinositol 4-phosphate (PI4P) and exchanged PS for PI4P between two membranes. We solved the crystal structure of Osh6p:PI4P complex and demonstrated that the transport of PS by Osh6p depends on PI4P recognition in vivo. Finally, we showed that the PI4P-phosphatase Sac1p, by maintaining a PI4P gradient at the ER/PM interface, drove PS transport. Thus, PS transport by oxysterol-binding protein-related protein (ORP)/oxysterol-binding homology (Osh) proteins is fueled by PI4P metabolism through PS/PI4P exchange cycles.


Subject(s)
Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Phosphatidylinositol Phosphates/metabolism , Phosphatidylserines/metabolism , Phosphoric Monoester Hydrolases/metabolism , Receptors, Steroid/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Biological Transport , Crystallography, X-Ray , Phosphatidylinositol Phosphates/chemistry , Phosphatidylserines/chemistry , Phosphoric Monoester Hydrolases/genetics , Receptors, Steroid/chemistry , Receptors, Steroid/genetics , Saccharomyces cerevisiae Proteins/genetics
10.
Article in English | MEDLINE | ID: mdl-26029163

ABSTRACT

Endocrine-disrupting chemicals (EDCs) are exogenous substances interfering with hormone biosynthesis, metabolism, or action, and consequently causing disturbances in the endocrine system. Various pathways are activated by EDCs, including interactions with nuclear receptors (NRs), which are primary targets of numerous environmental contaminants. The main NRs targeted by environmental contaminants are the estrogen (ER α, ß) and the androgen (AR) receptors. ERs and AR have pleiotropic regulatory roles in a diverse range of tissues, notably in the mammary gland, the uterus, and the prostate. Thus, dysfunctional ERs and AR signaling due to inappropriate exposure to environmental pollutants may lead to hormonal cancers and infertility. The pregnane X receptor (PXR) is also recognized by many environmental molecules. PXR has a protective role of the body through its ability to regulate proteins involved in the metabolism, the conjugation, and the transport of many exogenous and endogenous compounds. However, the permanent activation of this receptor by xenobiotics may lead to premature drug metabolism, the formation, and accumulation of toxic metabolites and defects in hormones homeostasis. The activity of other NRs can also be affected by environmental molecules. Compounds capable of inhibiting or activating the estrogen related (ERRγ), the thyroid hormone (TRα, ß), the retinoid X receptors (RXRα, ß, γ), and peroxisome proliferator-activated (PPAR α, γ) receptors have been identified and are highly suspected to promote developmental, reproductive, neurological, or metabolic diseases in humans and wildlife. In this review, we provide an overview of reporter cell lines established to characterize the human NR activities of a large panel of EDCs including natural as well as industrial compounds such as pesticides, plasticizers, surfactants, flame retardants, and cosmetics.

11.
Front Neurosci ; 9: 212, 2015.
Article in English | MEDLINE | ID: mdl-26106289

ABSTRACT

Zebrafish is increasingly used as an animal model to study the effects of environmental nuclear receptors (NRs) ligands. As most of these compounds have only been tested on human NRs, it is necessary to measure their effects on zebrafish NRs. Estrogen receptors (ER) α and ß and peroxysome proliferator activated receptor (PPAR) γ are main targets of environmental disrupting compounds (EDCs). In humans there are two distinct nuclear ERs (hERα and hERß), whereas the zebrafish genome encodes three ERs, zfERα, zfERß1, and zfERß2. Only one isoform of PPARγ is expressed in both humans and zebrafish. In this review, we described reporter cell lines that we established to study the interaction of EDCs with human and zebrafish ERs and PPARγ. Using these cell lines, we observed that zfERs are thermo-sensitive while zfPPARγ is not. We also showed significant differences in the ability of environmental and synthetic ligands to modulate activation of zfERs and zfPPARγ in comparison to hERs and hPPARγ. Some environmental estrogens (bisphenol A, mycoestrogens) which are hER panagonists displayed greater potency for zfERα as compared to zfERßs. hERß selective agonists (8ßVE2, DPN, phytoestrogens) also displayed zfERα selectivity. Among hERα selective synthetic agonists, 16α-LE2 was the most zfERα selective compound. Almost all zfPPARγ environmental ligands (halogenated bisphenol A derivatives, phthalates, perfluorinated compounds) displayed similar affinity for human and zebrafish PPARγ while pharmaceutical hPPARγ agonists like thiazolidones are not recognized by zfPPARγ. Altogether, our studies show that all hERs and hPPARγ ligands do not control in a similar manner the transcriptional activity of zfERs and zfPPARγ and point out that care has to be taken in transposing the results obtained using the zebrafish as a model for human physiopathology.

12.
PLoS One ; 10(5): e0123195, 2015.
Article in English | MEDLINE | ID: mdl-25933005

ABSTRACT

Retinoic acid is an important regulator of cell differentiation which plays major roles in embryonic development and tissue remodeling. The biological action of retinoic acid is mediated by three nuclear receptors denoted RARα, ß and γ. Multiple studies support that RARß possesses functional characteristics of a tumor suppressor and indeed, its expression is frequently lost in neoplastic tissues. However, it has been recently reported that RARß could also play a role in mammary gland tumorigenesis, thus demonstrating the important but yet incompletely understood function of this receptor in cancer development. As a consequence, there is a great need for RARß-selective agonists and antagonists as tools to facilitate the pharmacological analysis of this protein in vitro and in vivo as well as for potential therapeutic interventions. Here we provide experimental evidences that the novel synthetic retinoid BMS948 is an RARß-selective ligand exhibiting a full transcriptional agonistic activity and activating RARß as efficiently as the reference agonist TTNPB. In addition, we solved the crystal structures of the RARß ligand-binding domain in complex with BMS948 and two related compounds, BMS641 and BMS411. These structures provided a rationale to explain how a single retinoid can be at the same time an RARα antagonist and an RARß full agonist, and revealed the structural basis of partial agonism. Finally, in addition to revealing that a flip by 180° of the amide linker, that usually confers RARα selectivity, accounts for the RARß selectivity of BMS948, the structural analysis uncovers guidelines for the rational design of RARß-selective antagonists.


Subject(s)
Imidazoles/metabolism , Imidazoles/pharmacology , Receptors, Retinoic Acid/agonists , Receptors, Retinoic Acid/metabolism , Crystallography, X-Ray , HeLa Cells , Humans , Imidazoles/chemistry , Ligands , Models, Molecular , Protein Binding/drug effects , Protein Structure, Tertiary , Receptors, Retinoic Acid/chemistry , Retinoic Acid Receptor alpha , Structure-Activity Relationship
13.
Acta Pharmacol Sin ; 36(1): 88-101, 2015 01.
Article in English | MEDLINE | ID: mdl-25500867

ABSTRACT

Nuclear receptors (NRs) are members of a large superfamily of evolutionarily related transcription factors that control a plethora of biological processes. NRs orchestrate complex events such as development, organ homeostasis, metabolism, immune function, and reproduction. Approximately one-half of the 48 human NRs have been shown to act as ligand-regulated transcription factors and respond directly to a large variety of endogenous hormones and metabolites that are generally hydrophobic and small in size (eg, retinoic acid or estradiol). The second half of the NR family comprises the so-called orphan receptors, for which regulatory ligands are still unknown or may not exist despite the presence of a C-terminal ligand-binding domain, which is the hallmark of all NRs. Several chemicals released into the environment (eg, bisphenols, phthalates, parabens, etc) share some physicochemical properties with natural ligands, allowing them to bind to NRs and activate or inhibit their action. Collectively referred to as endocrine disruptors or endocrine-disrupting chemicals (EDCs), these environmental pollutants are highly suspected to cause a wide range of developmental, reproductive, neurological, or metabolic defects in humans and wildlife. Crystallographic studies are revealing unanticipated mechanisms by which chemically diverse EDCs interact with the ligand-binding domain of NRs. These studies thereby provide a rational basis for designing novel chemicals with lower impacts on human and animal health. In this review, we provide a structural and mechanistic view of endocrine disrupting action using estrogen receptors α and ß, (ERα/ß), peroxisome proliferator activated receptor γ (PPARγ), and their respective environmental ligands as representative examples.

14.
Environ Health Perspect ; 122(12): 1306-13, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25260197

ABSTRACT

BACKGROUND: Individuals are exposed daily to environmental pollutants that may act as endocrine-disrupting chemicals (EDCs), causing a range of developmental, reproductive, metabolic, or neoplastic diseases. With their mostly hydrophobic pocket that serves as a docking site for endogenous and exogenous ligands, nuclear receptors (NRs) can be primary targets of small molecule environmental contaminants. However, most of these compounds are chemically unrelated to natural hormones, so their binding modes and associated hormonal activities are hardly predictable. OBJECTIVES: We conducted a correlative analysis of structural and functional data to gain insight into the mechanisms by which 12 members of representative families of pollutants bind to and activate the estrogen receptors ERα and ERß. METHODS: We used a battery of biochemical, structural, biophysical, and cell-based approaches to characterize the interaction between ERs and their environmental ligands. RESULTS: Our study revealed that the chemically diverse compounds bound to ERs via varied sets of protein-ligand interactions, reflecting their differential activities, binding affinities, and specificities. We observed xenoestrogens binding to both ERs-with affinities ranging from subnanomolar to micromolar values-and acting in a subtype-dependent fashion as full agonists or partial agonists/antagonists by using different combinations of the activation functions 1 and 2 of ERα and ERß. CONCLUSIONS: The precise characterization of the interactions between major environmental pollutants and two of their primary biological targets provides rational guidelines for the design of safer chemicals, and will increase the accuracy and usefulness of structure-based computational methods, allowing for activity prediction of chemicals in risk assessment.


Subject(s)
Endocrine Disruptors/adverse effects , Environmental Pollutants/adverse effects , Receptors, Estrogen/metabolism , Endocrine Disruptors/chemistry , Environmental Exposure , Environmental Pollutants/chemistry , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , High-Throughput Screening Assays , Humans , Ligands , Structure-Activity Relationship
15.
Vitam Horm ; 94: 229-51, 2014.
Article in English | MEDLINE | ID: mdl-24388193

ABSTRACT

Bisphenol-A (BPA) is one of the highest-volume chemicals produced worldwide and the widespread exposure of individuals to BPA is suspected to affect a variety of physiological functions, including reproduction, development, and metabolism. Its estrogenic activity has been well documented in the last 15 years. In addition to estrogen receptors, BPA has been also shown to bind to and activate the estrogen-related receptor γ and pregnane X receptor and inhibit the androgen receptor. Halogenated BPAs were also shown to activate the peroxisome proliferator-activated receptor γ and inhibit thyroid hormone receptors. In this chapter, we review recent studies shedding light on the structural and molecular mechanisms by which BPA and its halogenated derivatives interfere with nuclear hormone receptor signaling. These data provide guidelines for the development of safer substitutes devoid of hormonal activity and may help environmental risk assessment.


Subject(s)
Androgen Antagonists/toxicity , Benzhydryl Compounds/toxicity , Environmental Pollutants/toxicity , Estrogens, Non-Steroidal/toxicity , PPAR gamma/agonists , Phenols/toxicity , Receptors, Steroid/agonists , Receptors, Thyroid Hormone/antagonists & inhibitors , Androgen Antagonists/chemistry , Animals , Benzhydryl Compounds/chemistry , Endocrine Disruptors/chemistry , Endocrine Disruptors/toxicity , Environmental Pollutants/chemistry , Estrogens, Non-Steroidal/chemistry , Halogenation , Humans , PPAR gamma/metabolism , Phenols/chemistry , Pregnane X Receptor , Receptors, Androgen/chemistry , Receptors, Androgen/metabolism , Receptors, Estrogen/agonists , Receptors, Estrogen/metabolism , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism
17.
Methods Mol Biol ; 941: 137-55, 2012.
Article in English | MEDLINE | ID: mdl-23065559

ABSTRACT

The increased awareness of the importance of RNA in biology, illustrated by the recent attention given to RNA interference research and applications, has spurred structural and functional investigations of RNA. For these studies, the traditional purification method for in vitro transcribed RNA is denaturing polyacrylamide gel electrophoresis. However, gel-based procedures denature the RNA and can be very tedious and time-consuming. Thus, several alternative schemes have been developed for fast non-denaturing purification of RNA transcribed in vitro. In a recent report, a quick affinity purification procedure was developed for RNAs transcribed with a 3'-ARiBo tag and shown to provide RNA with exceptionally high purity and yield. The ARiBo tag contains the λboxB RNA and the glmS ribozyme, allowing immobilization on GSH-Sepharose resin via a λN-GST fusion protein and elution by activation of the glmS ribozyme with glucosamine-6-phosphate. This Chapter outlines the experimental details for affinity batch purification of RNAs using ARiBo tags. Although the procedure was originally developed for purification of a stable purine riboswitch mutant, it is demonstrated here for purification of the terminal loop of the let-7g precursor miRNA, an important target of the pluripotency factor Lin28.


Subject(s)
Affinity Labels/metabolism , Bacteriophage lambda/genetics , Chromatography, Affinity/methods , RNA, Catalytic/metabolism , RNA, Viral/genetics , RNA/isolation & purification , RNA/metabolism , Base Sequence , Cloning, Molecular , Electrophoresis, Polyacrylamide Gel , Plasmids/genetics , RNA/chemistry , RNA Cleavage , Recombinant Fusion Proteins/metabolism , Transcription, Genetic
18.
Proc Natl Acad Sci U S A ; 109(37): 14930-5, 2012 Sep 11.
Article in English | MEDLINE | ID: mdl-22927406

ABSTRACT

Bisphenol A (BPA) is an industrial compound and a well known endocrine-disrupting chemical with estrogenic activity. The widespread exposure of individuals to BPA is suspected to affect a variety of physiological functions, including reproduction, development, and metabolism. Here we report that the mechanisms by which BPA and two congeners, bisphenol AF and bisphenol C (BPC), bind to and activate estrogen receptors (ER) α and ß differ from that used by 17ß-estradiol. We show that bisphenols act as partial agonists of ERs by activating the N-terminal activation function 1 regardless of their effect on the C-terminal activation function 2, which ranges from weak agonism (with BPA) to antagonism (with BPC). Crystallographic analysis of the interaction between bisphenols and ERs reveals two discrete binding modes, reflecting the different activities of compounds on ERs. BPA and 17ß-estradiol bind to ERs in a similar fashion, whereas, with a phenol ring pointing toward the activation helix H12, the orientation of BPC accounts for the marked antagonist character of this compound. Based on structural data, we developed a protocol for in silico evaluation of the interaction between bisphenols and ERs or other members of the nuclear hormone receptor family, such as estrogen-related receptor γ and androgen receptor, which are two known main targets of bisphenols. Overall, this study provides a wealth of tools and information that could be used for the development of BPA substitutes devoid of nuclear hormone receptor-mediated activity and more generally for environmental risk assessment.


Subject(s)
Endocrine Disruptors/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Models, Molecular , Phenols/metabolism , Animals , Benzhydryl Compounds , Cell Line , Chromatography, Gel , Crystallography , Dose-Response Relationship, Drug , Endocrine Disruptors/chemistry , Estradiol/metabolism , Estrogen Receptor alpha/isolation & purification , Estrogen Receptor beta/isolation & purification , Fluorescence Polarization , Humans , Luciferases , Oncorhynchus mykiss , Phenols/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Reverse Transcriptase Polymerase Chain Reaction
19.
J Cell Biol ; 195(6): 965-78, 2011 Dec 12.
Article in English | MEDLINE | ID: mdl-22162133

ABSTRACT

Osh/Orp proteins transport sterols between organelles and are involved in phosphoinositide metabolism. The link between these two aspects remains elusive. Using novel assays, we address the influence of membrane composition on the ability of Osh4p/Kes1p to extract, deliver, or transport dehydroergosterol (DHE). Surprisingly, phosphatidylinositol 4-phosphate (PI(4)P) specifically inhibited DHE extraction because PI(4)P was itself efficiently extracted by Osh4p. We solve the structure of the Osh4p-PI(4)P complex and reveal how Osh4p selectively substitutes PI(4)P for sterol. Last, we show that Osh4p quickly exchanges DHE for PI(4)P and, thereby, can transport these two lipids between membranes along opposite routes. These results suggest a model in which Osh4p transports sterol from the ER to late compartments pinpointed by PI(4)P and, in turn, transports PI(4)P backward. Coupled to PI(4)P metabolism, this transport cycle would create sterol gradients. Because the residues that recognize PI(4)P are conserved in Osh4p homologues, other Osh/Orp are potential sterol/phosphoinositol phosphate exchangers.


Subject(s)
Ergosterol/analogs & derivatives , Lipid Bilayers/chemistry , Membrane Proteins/chemistry , Phosphatidylinositol Phosphates/chemistry , Receptors, Steroid/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Ergosterol/chemistry , Liposomes/chemistry , Protein Structure, Tertiary
20.
Nucleic Acids Res ; 38(6): 2057-68, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20022916

ABSTRACT

The adenine and guanine riboswitches regulate gene expression in response to their purine ligand. X-ray structures of the aptamer moiety of these riboswitches are characterized by a compact fold in which the ligand forms a Watson-Crick base pair with residue 65. Phylogenetic analyses revealed a strict restriction at position 39 of the aptamer that prevents the G39-C65 and A39-U65 combinations, and mutational studies indicate that aptamers with these sequence combinations are impaired for ligand binding. In order to investigate the rationale for sequence conservation at residue 39, structural characterization of the U65C mutant from Bacillus subtilis pbuE adenine riboswitch aptamer was undertaken. NMR spectroscopy and X-ray crystallography studies demonstrate that the U65C mutant adopts a compact ligand-free structure, in which G39 occupies the ligand-binding site of purine riboswitch aptamers. These studies present a remarkable example of a mutant RNA aptamer that adopts a native-like fold by means of ligand mimicking and explain why this mutant is impaired for ligand binding. Furthermore, this work provides a specific insight into how the natural sequence has evolved through selection of nucleotide identities that contribute to formation of the ligand-bound state, but ensures that the ligand-free state remains in an active conformation.


Subject(s)
Adenine/chemistry , RNA, Bacterial/chemistry , Regulatory Sequences, Ribonucleic Acid , 5' Untranslated Regions , Adenine/metabolism , Aptamers, Nucleotide/chemistry , Bacillus subtilis/genetics , Base Sequence , Binding Sites , Crystallography, X-Ray , Guanine/chemistry , Guanine/metabolism , Ligands , Models, Molecular , Molecular Sequence Data , Mutation , Nuclear Magnetic Resonance, Biomolecular , Nucleic Acid Conformation , RNA, Bacterial/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...